WO2003020874A2 - Improved method for freezing viable cells - Google Patents

Improved method for freezing viable cells Download PDF

Info

Publication number
WO2003020874A2
WO2003020874A2 PCT/IL2002/000738 IL0200738W WO03020874A2 WO 2003020874 A2 WO2003020874 A2 WO 2003020874A2 IL 0200738 W IL0200738 W IL 0200738W WO 03020874 A2 WO03020874 A2 WO 03020874A2
Authority
WO
WIPO (PCT)
Prior art keywords
vessel
sample
freezing
cells
track
Prior art date
Application number
PCT/IL2002/000738
Other languages
French (fr)
Other versions
WO2003020874A3 (en
Inventor
Yehudit Natan
Meir Uri
Yoram Amsalem
Original Assignee
I.M.T Interface Multigrad Technology Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by I.M.T Interface Multigrad Technology Ltd. filed Critical I.M.T Interface Multigrad Technology Ltd.
Priority to US10/488,414 priority Critical patent/US7659111B2/en
Priority to AU2002337480A priority patent/AU2002337480A1/en
Priority to EP02772760A priority patent/EP1428008B1/en
Priority to AT02772760T priority patent/ATE542123T1/en
Publication of WO2003020874A2 publication Critical patent/WO2003020874A2/en
Publication of WO2003020874A3 publication Critical patent/WO2003020874A3/en

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L7/00Heating or cooling apparatus; Heat insulating devices
    • B01L7/54Heating or cooling apparatus; Heat insulating devices using spatial temperature gradients
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0236Mechanical aspects
    • A01N1/0242Apparatuses, i.e. devices used in the process of preservation of living parts, such as pumps, refrigeration devices or any other devices featuring moving parts and/or temperature controlling components
    • A01N1/0252Temperature controlling refrigerating apparatus, i.e. devices used to actively control the temperature of a designated internal volume, e.g. refrigerators, freeze-drying apparatus or liquid nitrogen baths
    • A01N1/0257Stationary or portable vessels generating cryogenic temperatures
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0278Physical preservation processes
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F25REFRIGERATION OR COOLING; COMBINED HEATING AND REFRIGERATION SYSTEMS; HEAT PUMP SYSTEMS; MANUFACTURE OR STORAGE OF ICE; LIQUEFACTION SOLIDIFICATION OF GASES
    • F25DREFRIGERATORS; COLD ROOMS; ICE-BOXES; COOLING OR FREEZING APPARATUS NOT OTHERWISE PROVIDED FOR
    • F25D3/00Devices using other cold materials; Devices using cold-storage bodies
    • F25D3/02Devices using other cold materials; Devices using cold-storage bodies using ice, e.g. ice-boxes
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F26DRYING
    • F26BDRYING SOLID MATERIALS OR OBJECTS BY REMOVING LIQUID THEREFROM
    • F26B5/00Drying solid materials or objects by processes not involving the application of heat
    • F26B5/04Drying solid materials or objects by processes not involving the application of heat by evaporation or sublimation of moisture under reduced pressure, e.g. in a vacuum
    • F26B5/06Drying solid materials or objects by processes not involving the application of heat by evaporation or sublimation of moisture under reduced pressure, e.g. in a vacuum the process involving freezing
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F25REFRIGERATION OR COOLING; COMBINED HEATING AND REFRIGERATION SYSTEMS; HEAT PUMP SYSTEMS; MANUFACTURE OR STORAGE OF ICE; LIQUEFACTION SOLIDIFICATION OF GASES
    • F25CPRODUCING, WORKING OR HANDLING ICE
    • F25C1/00Producing ice
    • F25C1/18Producing ice of a particular transparency or translucency, e.g. by injecting air
    • F25C1/20Producing ice of a particular transparency or translucency, e.g. by injecting air by agitation
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F25REFRIGERATION OR COOLING; COMBINED HEATING AND REFRIGERATION SYSTEMS; HEAT PUMP SYSTEMS; MANUFACTURE OR STORAGE OF ICE; LIQUEFACTION SOLIDIFICATION OF GASES
    • F25DREFRIGERATORS; COLD ROOMS; ICE-BOXES; COOLING OR FREEZING APPARATUS NOT OTHERWISE PROVIDED FOR
    • F25D3/00Devices using other cold materials; Devices using cold-storage bodies
    • F25D3/10Devices using other cold materials; Devices using cold-storage bodies using liquefied gases, e.g. liquid air
    • F25D3/11Devices using other cold materials; Devices using cold-storage bodies using liquefied gases, e.g. liquid air with conveyors carrying articles to be cooled through the cooling space
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/42Low-temperature sample treatment, e.g. cryofixation

Definitions

  • the present invention relates to the storage of living cells and in particular relates to a method of. and device for, freezing living cells.
  • the method can be applied to freeze high volume samples or for the freezing as a preparatory step preceding lyophilization.
  • the cells be held in a state of suspended animation, a state wherein cell metabolism is entirely stopped. When needed, the cells are reanimated and used. Cells such as spermatozoa, blood and blood products are amongst the cell types for which long term storage is desired.
  • a generally accepted way of storing cells in a state of suspended animation is cryopreservation. Most simply, a sample containing viable cells is quickly cooled, for example by immersion in liquid nitrogen, to freeze the sample. When thawed under carefully controlled conditions acceptable levels of post-thaw cell viability are achieved.
  • cryopreservation of cells involves the difficulties in maintaining the frozen state. Refrigeration at cryogenic temperatures of 198K is necessarily expensive (excepting in the Antarctic) and not a perfect solution as biological activity does not cease until about 143K. The bulk of the required refrigeration systems renders cryopreserved cells not easily transportable.
  • An additional method for storing cells in a state of suspended animation involves removing a significant proportion of water from the cells (drying).
  • dry cells can potentially be stored at close to room temperatures without requiring bulky and expensive storage devices.
  • cells are air-dried: the cells in solution are spread out in a thin layer and water allowed to evaporate.
  • air drying has not been shown to be a practical method for the preservation of large-volume samples. Post-rehydration viability of air-dried cells is incidental at best. Apparently, either the air-drying process or the rehydration process causes cytolysis.
  • Another approach for drying cells involves freeze-drying.
  • the simplest methods involve lyophilization of a frozen sample. Post-rehydration viability is poor.
  • cryoprotectants are antifreeze molecules and includes dimethyl sulfoxide (DMSO), glycerol and related polyhydril alcohols (e.g. propylene or ethylene glycol).
  • DMSO dimethyl sulfoxide
  • glycerol glycerol
  • polyhydril alcohols e.g. propylene or ethylene glycol
  • cryoprotectants termed non-permeating or extracellular cryoprotectants, bind to membrane lipids.
  • the membrane lipids are stabilized, thus preserving cell-structure integrity.
  • cryoprotectants include proline and sugars (e.g. glucose). Crowe (in U.S. 4,857,319) has taught that trehalose is an exceptionally good cryoprotectant.
  • cryoprotectants are manifold. Different cell types require different cryoprotectants at different concentrations. Thus the use of cryoprotectants is not a general method but rather requires a complex and time-consuming optimization step. Second, rehydration and thawing must be very carefully due to the effect of osmotic pressure resulting from the presence of cryoprotectants in solution. Lastly, upon rehydration or thawing, the cells are found in an "unnatural" solution which must be purified or modified before being used.
  • FIG. 1 Schematically depicted in Figure 1 is a device 10 used to realize the teachings of U.S. 5,873,254.
  • a vessel 12 holding a biological sample 14 lays on a track 16.
  • Track 16 leads from the surroundings into a cooling device 18.
  • Cooling device 18 is substantially a block of a thermally conductive material having a tunnel 20 through which vessel 12 can pass .when guided along track 16.
  • In cooling device 18 is maintained a temperature gradient by the use of a plurality of cooling elements 22a, 22b and 22c. The temperature gradient is oriented so that there is a temperature gradient parallel to tunnel 20.
  • the temperature of cooling device 18 at entrance 24 of tunnel 20 is typically roughly the temperature of sample 14 at the beginning of the freezing process. Exit 26 of tunnel 20 is cooler than entrance 24.
  • a toothed rod 28, a gear 30, configured to engage rod 28, and an electrical motor 32, configured to rotate gear 30, are all elements of a mechanism configured to push vessel 12 into and through tunnel 20 at a controlled rate.
  • the cooling rate C (in units of °C/min) is determined by the temperature gradient G (in units of °C/mm) along track 16 and the rate of advancement of the cooling front P (in units of mm/sec) through biological sample 14, determined by the speed which vessel 12 travels through tunnel 20.
  • Cells in cryoprotectant-containing samples have exceptional post-thaw viability when frozen according to the method of U.S. 5,873,254, for example by using a device such as device 10. It is important to note that when a biological sample is lyophilized subsequent to freezing according to the method of U.S. 5,873,254, post-rehydration cell viability is insufficient.
  • the method of the present invention involves directional cooling of a biological sample, substantially as described hereinabove and in U.S. 5,873,254, but unique in that as biological sample containing vessel advances through the thermal gradient, the vessel is agitated.
  • the preferred agitation mode is rotation, the axis of rotation preferably being substantially perpendicular to the direction of advancement.
  • a method for freezing cells including confining a sample in a vessel (the vessel having a leading end and a terminal end, the sample containing the cells and having a freezing point) and gradually cooling the sample to below the freezing point so as to generate a temperature gradient in the sample, where during cooling the leading end is cooler than the terminal end so as to generate a freezing front moving from the leading end towards the terminal end where during the gradual cooling the vessel is agitated.
  • the agitation is rotation of the vessel about an axis, the axis being substantially perpendicular to the freezing front.
  • the vessel is not completely filled with sample, preferably being less than about 90% filled, more preferably less than about 50% filled, and even more preferably less than about 30% filled.
  • the partial filling of the vessel by sample leads to a frozen sample with a large surface area and a crystal structure that allows for simple lyophilization and for exceptional post-rehydration viability of thus freeze-dried cells.
  • a method for storing viable freeze dried cells by freeze-drying a sample containing the cells as described above and subsequently storing the sample at a storage temperature greater than about 193K, greater than about 253K and even greater than about 273K for a period of time greater than 1 day, greater than one week and even greater than 6 weeks.
  • cryoprotectant that is solid at the storage temperature.
  • cryoprotectant is trehalose.
  • the device of the present invention is substantially a device for directional cooling of biological samples as described in U.S. 5,873,254 but additionally including a means of agitating the vessel in which the biological sample to be frozen is contained.
  • Agitation of the sample can be performed in a large number of ways. Conceivably shaking, vibrating, rocking, rolling can all be implemented as agitation methods to ensure that the biological sample is mixed during the freezing process and as a result the thermal homogeneity in the cooling front. Simplest to implement is rotation of the vessel, especially when the axis of rotation is parallel to the direction of advancement and thus perpendicular to the cooling front. Intuitively it seems that rotation perpendicular to the cooling front is the agitation method that allows for the greatest thermal homogeneity within the cooling front without disrupting the temperature gradient within the biological sample.
  • the word vessel as used herein is non-limiting and refers to a vessel in the usual sense of the word and includes such objects as bags, bottles, flasks, jars, tubes, boxes, sacks and other containers. While not wishing to be held to any one theory, it is believed that the agitation of the sample during directional cooling modifies the thermal homogeneity during the freezing process as well as the crystal morphology. Less cell damage occurs during the freezing process. When the sample does not entirely fill the test tube, the total surface area of the frozen sample is increased, allowing quick and efficient lyophilization without damaging cell membranes.
  • a device for freezing a sample including a vessel for containing the sample, a mechanism for moving said vessel along a track and a refrigeration means for imposing a laterally variable temperature gradient along the track, the refrigeration means including a plurality of thermally conductive blocks substantially- enclosing the track
  • the device also includes a a means of agitating the vessel when moving along the track.
  • the vessel is a tube with a diameter of greater than about 6 mm.
  • the means of agitating the vessel includes a motor, such as an electrical motor.
  • the means of agitating the ⁇ 'essel includes a control device to vary the intensity of agitation of the vessel.
  • the means of agitating the vessel are in fact means of rotating the vessel.
  • rotations is around an axis substantially parallel to the track, that is substantially perpendicular to a cold front generated inside the sample when the device is operated (vide infra).
  • the rotation is caused by a motor, such as an electrical motor.
  • the device of the present invention is fitted with a control device to vary the rate of rotation of the vessel.
  • FIGS. 1 are schematic depictions of a directional cooling device
  • FIG. 2 is a schematic depiction of a cooling device of the present invention.
  • the present invention is directed to a method and a device for the freezing of cells in solution so that the frozen cells can be lyophilized, stored and subsequently rehydrated.
  • the principles and uses of the method according to the present invention may be better understood with reference to description, the experimental results and the drawings, in which like reference numerals refer to like parts throughout all of the figures. It is understood that the descriptions herein are illustrative and not intended to restrict the present invention to the specific details set forth below.
  • FIG 2 is depicted a cooling device 50 of the present invention.
  • Clearly device 50 is similar to device 10 depicted in Figures 1.
  • rod 28 is tipped with engaging tines 54 and can be made to rotate around an axis parallel to track 16 by electrical rotation motor 52.
  • the speed by which rotation motor 52 rotates rod 28 is determined by rotation controller 56;
  • Engaging tines 54 are configured to engage following end 36 of vessel 12 when vessel 12 is on track 16.
  • the method of the present invention When the method of the present invention is used to freeze a sample, it is not necessary to add cryoprotectant to the sample. For a given sample, freezing according to the method of the present invention gives a significant improvement of post-thawing, and if lyophilized, post-rehydration cell viability when compared to freezing using the teachings of U.S. 5,873,254. That said, the method of the present invention is compatible with the use of cryoprotectants when and if desired.
  • large volumes of biological sample can be prepared for cryogenic storage at one time, a significant improvement of the teachings of U.S. 5,873,254.
  • the much greater volumes that can be frozen are a result of the fact that agitation of the sample-containing vessel during the freezing process allows greater thermal homogeneity within the sample, allowing a vessel with a significantly greater diameter to be used.
  • the largest practical vessel when using the teachings of U.S. 5,873,254 is about 5 cm long with a diameter of no more than about 0.5 cm.
  • vessels with diameters of greater than 6 mm, and even diameters of greater than 25 mm are typically used with no statistically significant reduction of post-rehydration viability.
  • Increased sample volume coupled with a faster cooling rate described above allows for a significant throughput increase.
  • freezing is a preparatory step for lyophilization.
  • the vessel not be filled completely with biological sample.
  • the biological sample fills only 90% of the vessel, more preferably only 50% of the vessel and most preferably only 30% of the vessel is filled.
  • the frozen sample so produced has a very high surface area. The high surface area, as well as the fact that no cryoprotectant needs be added, means that subsequent lyophilization is quick and efficient. A combination of a high freezing-rate, high sample volume, and fast lyophilization allows for a significant throughput increase.
  • a sample lyophilized after being frozen according to the method of the present invention can be stored at relatively high temperatures for extended periods of time without significant post-hydration viability loss.
  • In general storage temperature depends on the water loss. If a sample loses up to about 90% of the original weight due to lyophilization, the sample is stored at temperatures below freezing. If a sample loses more than about 90% of the original weight due to lyophilization, it may be possible to store the sample at high temperatures (277K or even room temperature).
  • Leukocytes were separated from whole blood using Histopaq ⁇ e 1077. The samples were put in standard 15ml test tubes and centrifuged for 30 minutes at 1000 G. The leukocyte layer was transferred to a different test tube and again centrifuged for 10 minutes at 250 G. The supernatant was discarded and the leukocyte pellet was washed with PBS and again centrifuged, as described above.
  • a leukocyte suspension was prepared by dissolving the leukocyte pellet in a solution containing 50% PBS (phosphate buffered solution) and 50% plasma. Trehalose was added so as to get a 0.1M trehalose concentration.
  • PBS phosphate buffered solution
  • 2.8 ml of the leukocyte suspension was transferred to each of three 12 ml glass test tube and frozen in a device of the present invention, a modified (resembling device 50 depicted in Figure 2) MTG freezing apparatus manufactured by IMT, Israel.
  • the thermal gradient in the cooling unit was set to 0.43°C/mm in all three cases with a final temperature of 203K.
  • the rate of test tube insertion was 0.02, 0.2 and 2.0 mm/sec, yielding cooling rates of 0.5, 5.1 and 51 °C/min, respectively.
  • the test tubes were rotated at a rate of 32 rpm.
  • test tube was taken from each test tube to test for cell viability, and the test tubes transferred to a lyophilizer. Lyophilization was performed at a chamber temperature of 238K and a condenser temperature of 191K for 3 hours and for 24 hours. A sample was taken from each test tube to test for viability after 3 hours lyophilization and after 24 hours lyophilization. After 24 hours of lyophilization test tube A lost 76% of the original sample weight, test tube B lost 77% of the original sample weight and sample C lost 87% of the original sample weight.
  • Viability %; test tube Interface Cooling weight sample 1 sample 2 sample 3 velocity rate loss (freeze/thaw) lyophilize lyophilize
  • test tubes D, E, F and G were prepared as above to examine the influence of a different interface velocity at a cooling rate of 5.1 °C / min cases to a final temperature of 253K, 238K, 203K, 173K, respectively. Samples were taken at intervals as described above. The test tubes were lyophilized to complete dryness (loss of greater than 92% of the initial weight) by lyophilization for 72 hours. After 72 hours lyophilization an additional sample was taken from each test tube. The results are presented in Table 2.
  • test tube Interface sample 1 sample 2 sample 3 sample 4 velocity (freeze/thaw) lyophilize lyophilize lyophilize lyophilize
  • test tubes FI, I and J Six further identical test tubes were prepared as above to examine the influence of a different rotation velocities at a cooling rate of 5.1 °C / min and an interface velocity of 0.2 mm/sec (test tubes FI, I and J) to a final temperature of 203K and 0.48 mm/sec (test tubes K,
  • test tube rotation sample 1 sample 2 sample 3 sample 4 rate (freeze/thaw) lyophilize lyophilize lyophilize lyophilize

Abstract

A method and a device (50) for improved freezing of biological samples containing living cells is disclosed. Directional cooling coupled with agitation, preferably rotation, of the vessel (12) containing the biological sample (14) allows for the freezing of large volume samples at much greater rates than before. Whether a sample is large or not, the greater thermal homogeneity during the freezing process as well as the resulting crystal morphology yield a frozen sample which is exceptionally suited for lyophilization.

Description

IMPROVED METHOD FOR FREEZING VIABLE CELLS
FIELD OF THE INVENTION
The present invention relates to the storage of living cells and in particular relates to a method of. and device for, freezing living cells. The method can be applied to freeze high volume samples or for the freezing as a preparatory step preceding lyophilization.
BACKGROUND OF THE INVENTION
There is often a need to store viable cells for extended periods of time. It is most preferred that the cells be held in a state of suspended animation, a state wherein cell metabolism is entirely stopped. When needed, the cells are reanimated and used. Cells such as spermatozoa, blood and blood products are amongst the cell types for which long term storage is desired.
A generally accepted way of storing cells in a state of suspended animation is cryopreservation. Most simply, a sample containing viable cells is quickly cooled, for example by immersion in liquid nitrogen, to freeze the sample. When thawed under carefully controlled conditions acceptable levels of post-thaw cell viability are achieved.
The greatest practical disadvantage of cryopreservation of cells involves the difficulties in maintaining the frozen state. Refrigeration at cryogenic temperatures of 198K is necessarily expensive (excepting in the Antarctic) and not a perfect solution as biological activity does not cease until about 143K. The bulk of the required refrigeration systems renders cryopreserved cells not easily transportable.
An additional method for storing cells in a state of suspended animation involves removing a significant proportion of water from the cells (drying). The greatest advantage of drying cells is that dry cells can potentially be stored at close to room temperatures without requiring bulky and expensive storage devices. Typically, cells are air-dried: the cells in solution are spread out in a thin layer and water allowed to evaporate. Although clearly there is no danger of ice crystal formation damaging the cells, the fact that the drying process occurs at a temperature of at least above 273K where cell metabolism is active is a great disadvantage. Further, air drying has not been shown to be a practical method for the preservation of large-volume samples. Post-rehydration viability of air-dried cells is incidental at best. Apparently, either the air-drying process or the rehydration process causes cytolysis.
A number of researchers have found methods of improving post-rehydration viability of air-dried cells by the addition of cell membrane stabilizers. Such stabilizers added to a buffered cell-containing solution improve post-rehydration viability.
Another approach for drying cells involves freeze-drying. The simplest methods involve lyophilization of a frozen sample. Post-rehydration viability is poor.
It has been found that an effective methods of increasing post-thaw or post-rehydration viability of frozen or freeze-dried cells, respectively, is the addition of cryoprotectants, substances that protect cell membranes from the effects of freezing.
One group of cryoprotectants. termed permeating cryoprotectants are antifreeze molecules and includes dimethyl sulfoxide (DMSO), glycerol and related polyhydril alcohols (e.g. propylene or ethylene glycol).
Another group of cryoprotectants termed non-permeating or extracellular cryoprotectants, bind to membrane lipids. The membrane lipids are stabilized, thus preserving cell-structure integrity. Such cryoprotectants include proline and sugars (e.g. glucose). Crowe (in U.S. 4,857,319) has taught that trehalose is an exceptionally good cryoprotectant.
The disadvantages of using cryoprotectants are manifold. Different cell types require different cryoprotectants at different concentrations. Thus the use of cryoprotectants is not a general method but rather requires a complex and time-consuming optimization step. Second, rehydration and thawing must be very carefully due to the effect of osmotic pressure resulting from the presence of cryoprotectants in solution. Lastly, upon rehydration or thawing, the cells are found in an "unnatural" solution which must be purified or modified before being used.
Kusakabe el al. ("Maintenance of genetic integrity in frozen and freeze-dried mouse spermatozoa" Proc. Nail. Acad. Sci... U.S.A. November 20 2001, 98(24), 13501-13506) has convincingly demonstrated that freeze-drying is a potentially successful strategy for the long term storage of cells. Mouse spermatozoa in a buffered solution without cryoprotectants were freeze dried by immersion in liquid nitrogen followed by 4 hours lyophilization. The freeze dried samples were stored at 277K for up to 56 days. The results indicate a high level of DNA integrity after rehydration. However, the usefulness of the teachings of Kusakabe et al. is limited by the fact that the cells were lyophilized in 0.1 ml portions. One skilled in the art would remain unconvinced of the potential of this method for the preservation of cells on a larger scale.
In U.S. 5,873.254. which is incorporated by reference for all purposes as if fully set forth herein, is described an innovative method for the freezing of biological samples by directional cooling. Schematically depicted in Figure 1 is a device 10 used to realize the teachings of U.S. 5,873,254. In Figure 1 A, a vessel 12 holding a biological sample 14 lays on a track 16. Track 16 leads from the surroundings into a cooling device 18. Cooling device 18 is substantially a block of a thermally conductive material having a tunnel 20 through which vessel 12 can pass .when guided along track 16. In cooling device 18 is maintained a temperature gradient by the use of a plurality of cooling elements 22a, 22b and 22c. The temperature gradient is oriented so that there is a temperature gradient parallel to tunnel 20. The temperature of cooling device 18 at entrance 24 of tunnel 20 is typically roughly the temperature of sample 14 at the beginning of the freezing process. Exit 26 of tunnel 20 is cooler than entrance 24. A toothed rod 28, a gear 30, configured to engage rod 28, and an electrical motor 32, configured to rotate gear 30, are all elements of a mechanism configured to push vessel 12 into and through tunnel 20 at a controlled rate.
When it is desired to freeze biological sample 14, motor 32 is activated causing rod 28 to push vessel 12 into cooling device 18 through tunnel 20, Figure IB. Throughout the travel of biological sample 14 through tunnel 20, each portion of biological sample is cooled at a similar rate, but at any given moment the temperature of biological sample near the leading end 34 of vessel 12 is lower then that of biological sample near the following end 36 of vessel 12. A cooling front is formed inside biological sample 14, the cooling front traveling from leading end 34 towards following end 36, in parallel to the motion of vessel 12 along track 16. Biological sample 14 found in a given cross-section perpendicular to the motion of vessel 12 along track 16 is substantially homogenous in temperature. Through biological sample 14 is a temperature gradient parallel to track 16, the temperature gradient substantially mirroring the temperature gradient found along track 16.
Practically, it has been found that to prevent supercooling of biological sample 14 (and the concomitant uncontrolled and inhomogeneous freezing rate, see U.S. 5,873,254) it is necessary to "seed" biological sample near leading end 34 before entering tunnel 20. Seeding is done, for example, by applying liquid nitrogen to leading end 34.
An important parameter when applying the method of U.S. 5,873,254 is the cooling rate. The cooling rate C (in units of °C/min) is determined by the temperature gradient G (in units of °C/mm) along track 16 and the rate of advancement of the cooling front P (in units of mm/sec) through biological sample 14, determined by the speed which vessel 12 travels through tunnel 20.
Cells in cryoprotectant-containing samples have exceptional post-thaw viability when frozen according to the method of U.S. 5,873,254, for example by using a device such as device 10. It is important to note that when a biological sample is lyophilized subsequent to freezing according to the method of U.S. 5,873,254, post-rehydration cell viability is insufficient.
A practical disadvantage of the method of U.S. 5,873,254 arises from the relatively limited amounts of sample that can be frozen at any one time. Experimentally it has been found that post-rehydration viability decreases when large volumes (generally greater than 1 ml) are frozen.
It would be highly advantageous to have a method of freezing large volume samp]es containing living cells. It would be exceptionally advantageous if such a method froze the sample in a way as to allow high post-rehydration post-lyophilization viability. It would be advantageous if such a method did not require the use of cryoprotectants.
SUMMARY OF THE INVENTION
The above and other objectives are achieved by the innovative method of freezing biological samples and use of the innovative device provided by the present invention.
The method of the present invention involves directional cooling of a biological sample, substantially as described hereinabove and in U.S. 5,873,254, but unique in that as biological sample containing vessel advances through the thermal gradient, the vessel is agitated. The preferred agitation mode is rotation, the axis of rotation preferably being substantially perpendicular to the direction of advancement.
According to the teachings of the present invention there is provided a method for freezing cells including confining a sample in a vessel (the vessel having a leading end and a terminal end, the sample containing the cells and having a freezing point) and gradually cooling the sample to below the freezing point so as to generate a temperature gradient in the sample, where during cooling the leading end is cooler than the terminal end so as to generate a freezing front moving from the leading end towards the terminal end where during the gradual cooling the vessel is agitated. According to a feature of the present invention the agitation is rotation of the vessel about an axis, the axis being substantially perpendicular to the freezing front.
According to a feature of the present invention the vessel is not completely filled with sample, preferably being less than about 90% filled, more preferably less than about 50% filled, and even more preferably less than about 30% filled. The partial filling of the vessel by sample leads to a frozen sample with a large surface area and a crystal structure that allows for simple lyophilization and for exceptional post-rehydration viability of thus freeze-dried cells.
According to the teachings of the present invention there is provided a method for freeze-drying cells by freezing a sample containing the cells as described above and subsequently lyophilizing the frozen sample.
According to the teachings of the present invention there is provided a method for storing viable freeze dried cells by freeze-drying a sample containing the cells as described above and subsequently storing the sample at a storage temperature greater than about 193K, greater than about 253K and even greater than about 273K for a period of time greater than 1 day, greater than one week and even greater than 6 weeks.
According to a feature of the present invention to the sample, before freeze-drying is added a cryoprotectant, that is solid at the storage temperature. Preferably the cryoprotectant is trehalose.
The device of the present invention is substantially a device for directional cooling of biological samples as described in U.S. 5,873,254 but additionally including a means of agitating the vessel in which the biological sample to be frozen is contained.
Agitation of the sample can be performed in a large number of ways. Conceivably shaking, vibrating, rocking, rolling can all be implemented as agitation methods to ensure that the biological sample is mixed during the freezing process and as a result the thermal homogeneity in the cooling front. Simplest to implement is rotation of the vessel, especially when the axis of rotation is parallel to the direction of advancement and thus perpendicular to the cooling front. Intuitively it seems that rotation perpendicular to the cooling front is the agitation method that allows for the greatest thermal homogeneity within the cooling front without disrupting the temperature gradient within the biological sample.
The word vessel as used herein is non-limiting and refers to a vessel in the usual sense of the word and includes such objects as bags, bottles, flasks, jars, tubes, boxes, sacks and other containers. While not wishing to be held to any one theory, it is believed that the agitation of the sample during directional cooling modifies the thermal homogeneity during the freezing process as well as the crystal morphology. Less cell damage occurs during the freezing process. When the sample does not entirely fill the test tube, the total surface area of the frozen sample is increased, allowing quick and efficient lyophilization without damaging cell membranes.
Thus according to the teachings of the present invention is provided a device for freezing a sample (especially a living cell containing sample of biological origin) including a vessel for containing the sample, a mechanism for moving said vessel along a track and a refrigeration means for imposing a laterally variable temperature gradient along the track, the refrigeration means including a plurality of thermally conductive blocks substantially- enclosing the track where the device also includes a a means of agitating the vessel when moving along the track.
According to a feature of the device of the present invention, the vessel is a tube with a diameter of greater than about 6 mm.
According to a further feature of the device of the present invention the means of agitating the vessel includes a motor, such as an electrical motor. According to a feature of the device of the present invention the means of agitating the λ'essel includes a control device to vary the intensity of agitation of the vessel.
According to a still further feature of the present invention, the means of agitating the vessel are in fact means of rotating the vessel. Preferably, rotations is around an axis substantially parallel to the track, that is substantially perpendicular to a cold front generated inside the sample when the device is operated (vide infra). According to a feature of the present invention the rotation is caused by a motor, such as an electrical motor. Preferably the device of the present invention is fitted with a control device to vary the rate of rotation of the vessel.
It is important to note that the method and device of the present invention are developed for and described herein as relates to samples of a biological origin containing living cells. However as is clear to one skilled in the art, the teachings of the present invention can be applied to and include other complex structures that are susceptible to freeze-damage such as (but not limited to) proteins, nucleic acids, liposomes, micelles, vesicles, cells, viruses, organs and organisms. BRIEF DESCRIPTION OF THE DRAWINGS
The invention is herein described, by way of example only, with reference to the accompanying drawings, wherein:
FIGS. 1 (prior art) are schematic depictions of a directional cooling device; and
FIG. 2 is a schematic depiction of a cooling device of the present invention.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to a method and a device for the freezing of cells in solution so that the frozen cells can be lyophilized, stored and subsequently rehydrated. The principles and uses of the method according to the present invention may be better understood with reference to description, the experimental results and the drawings, in which like reference numerals refer to like parts throughout all of the figures. It is understood that the descriptions herein are illustrative and not intended to restrict the present invention to the specific details set forth below.
In Figure 2 is depicted a cooling device 50 of the present invention. Clearly device 50 is similar to device 10 depicted in Figures 1. However, unlike in device 10, in device 50 rod 28 is tipped with engaging tines 54 and can be made to rotate around an axis parallel to track 16 by electrical rotation motor 52. The speed by which rotation motor 52 rotates rod 28 is determined by rotation controller 56; Engaging tines 54 are configured to engage following end 36 of vessel 12 when vessel 12 is on track 16.
When motor 32 is activated to cause rod 28 to push vessel 12 into tunnel 20, rotation motor 52 is also activated so as to rotate rod 28, engaging tines 54 and consequently vessel 12.
When the method of the present invention is used to freeze a sample, it is not necessary to add cryoprotectant to the sample. For a given sample, freezing according to the method of the present invention gives a significant improvement of post-thawing, and if lyophilized, post-rehydration cell viability when compared to freezing using the teachings of U.S. 5,873,254. That said, the method of the present invention is compatible with the use of cryoprotectants when and if desired.
In addition, due to the increased thermal homogeneity achieved by agitation of the sample-containing vessel, a much greater cooling rate (greater than 1°C / min and even greater than 50°C / min) can be used without reducing post-thawing or post-rehydration viability. The advantages of the present invention can best be understood by considering two specific embodiments of the method of the present invention: freezing large volumes of a biological sample at one time and freezing biological samples as a step preceding lyophilization for long term storage of viable cells.
In a first embodiment of the present invention, large volumes of biological sample can be prepared for cryogenic storage at one time, a significant improvement of the teachings of U.S. 5,873,254. The much greater volumes that can be frozen are a result of the fact that agitation of the sample-containing vessel during the freezing process allows greater thermal homogeneity within the sample, allowing a vessel with a significantly greater diameter to be used. Experiments show that the largest practical vessel when using the teachings of U.S. 5,873,254 is about 5 cm long with a diameter of no more than about 0.5 cm. When using the teachings of the present invention, vessels with diameters of greater than 6 mm, and even diameters of greater than 25 mm, are typically used with no statistically significant reduction of post-rehydration viability. Increased sample volume coupled with a faster cooling rate described above allows for a significant throughput increase.
In a second embodiment of the present invention, freezing is a preparatory step for lyophilization. According to the present invention, when a' sample is prepared for lyophilization, it is preferred that the vessel not be filled completely with biological sample. Preferred is that the biological sample fills only 90% of the vessel, more preferably only 50% of the vessel and most preferably only 30% of the vessel is filled. The frozen sample so produced has a very high surface area. The high surface area, as well as the fact that no cryoprotectant needs be added, means that subsequent lyophilization is quick and efficient. A combination of a high freezing-rate, high sample volume, and fast lyophilization allows for a significant throughput increase. More importantly, a sample lyophilized after being frozen according to the method of the present invention can be stored at relatively high temperatures for extended periods of time without significant post-hydration viability loss. In general storage temperature depends on the water loss. If a sample loses up to about 90% of the original weight due to lyophilization, the sample is stored at temperatures below freezing. If a sample loses more than about 90% of the original weight due to lyophilization, it may be possible to store the sample at high temperatures (277K or even room temperature).
Rehydration of a sample lyophilized according to the method of the present invention is simple. After the sample is thawed in the usual way, a suitable liquid such as water is added. EXPERIMENTAL RESULTS
Unless otherwise noted, all materials were purchased from Sigma Inc. (St. Louis, Missouri, USA).
Freezing and Lyophilization of Leukocytes
Leukocytes were separated from whole blood using Histopaqπe 1077. The samples were put in standard 15ml test tubes and centrifuged for 30 minutes at 1000 G. The leukocyte layer was transferred to a different test tube and again centrifuged for 10 minutes at 250 G. The supernatant was discarded and the leukocyte pellet was washed with PBS and again centrifuged, as described above.
A leukocyte suspension was prepared by dissolving the leukocyte pellet in a solution containing 50% PBS (phosphate buffered solution) and 50% plasma. Trehalose was added so as to get a 0.1M trehalose concentration.
2.8 ml of the leukocyte suspension was transferred to each of three 12 ml glass test tube and frozen in a device of the present invention, a modified (resembling device 50 depicted in Figure 2) MTG freezing apparatus manufactured by IMT, Israel. The thermal gradient in the cooling unit was set to 0.43°C/mm in all three cases with a final temperature of 203K. The rate of test tube insertion (interface velocity) was 0.02, 0.2 and 2.0 mm/sec, yielding cooling rates of 0.5, 5.1 and 51 °C/min, respectively. During insertion the test tubes were rotated at a rate of 32 rpm. Subsequent to freezing, a sample was taken from each test tube to test for cell viability, and the test tubes transferred to a lyophilizer. Lyophilization was performed at a chamber temperature of 238K and a condenser temperature of 191K for 3 hours and for 24 hours. A sample was taken from each test tube to test for viability after 3 hours lyophilization and after 24 hours lyophilization. After 24 hours of lyophilization test tube A lost 76% of the original sample weight, test tube B lost 77% of the original sample weight and sample C lost 87% of the original sample weight.
Each of the nine samples taken was allowed to thaw at room temperature and the cells rehydrated with distilled water. Viability was assessed using SYBR14/propidium iodide fluorescent staining (Molecular Probes Inc., Eugene, Oregon, USA). The viability results are summarized in Table 1. TABLE 1
Viability (%; test tube Interface Cooling weight sample 1 sample 2 sample 3 velocity rate loss (freeze/thaw) lyophilize lyophilize
[mm/sec] [°C / min] (%) (3h) (24h)
A 0.02 0.5 76 59 38 30 B 0.2 5.1 77 78 48 47 C 2 51 87 71 30 46
From the results presented in Table 1 it becomes clear that when it is desired to freeze-dry a large volume sample (in this case 2.8 ml) and preserve post-thaλv λάability, the method of the present invention gives remarkable results. Also seen is that the ideal interface velocity is very fast, indicating the usefulness of the present invention in a high throughput environment.
Four further identical test tubes, D, E, F and G were prepared as above to examine the influence of a different interface velocity at a cooling rate of 5.1 °C / min cases to a final temperature of 253K, 238K, 203K, 173K, respectively. Samples were taken at intervals as described above. The test tubes were lyophilized to complete dryness (loss of greater than 92% of the initial weight) by lyophilization for 72 hours. After 72 hours lyophilization an additional sample was taken from each test tube. The results are presented in Table 2.
TABLE 2
Viability (%) test tube Interface sample 1 sample 2 sample 3 sample 4 velocity (freeze/thaw) lyophilize lyophilize lyophilize
[mm sec] (3h) (24h) (72h)
D 0.13 76 74 22 7
E 0.2 75 62 38 12
F 0.48 65 55 35 8
G 1.0 76 49 22 6
From the results in Table 2 it is seen that a high post-rehydration viability can be expected of cells dried according to the method of the present invention Six further identical test tubes were prepared as above to examine the influence of a different rotation velocities at a cooling rate of 5.1 °C / min and an interface velocity of 0.2 mm/sec (test tubes FI, I and J) to a final temperature of 203K and 0.48 mm/sec (test tubes K,
L and M) to a final temperature of 238K. Samples were taken at intervals as described above.
The results are presented in Table 3.
TABLE 3
Viability (%) test tube rotation sample 1 sample 2 sample 3 sample 4 rate (freeze/thaw) lyophilize lyophilize lyophilize
[rpm] (3h) (24h) ' (72h)
jly - 0.22 mm/sec
H 1 85 60 38 18
I 32 72 48 . 40 22
J 140 76 52 19 23 ity = 0.48 mm/sec
K 1 72 46 25 14 L 32 62 53 33 6
M 140 70 44 18 18
From the results in Table 3 it is seen that very good viability can be expected of completely dried cells. It is seen that when rotation is relatively fast, there is less viability loss between partial and complete drying, presumably due to better thermal homogeneity during freezing and drying.
While the invention has been described with respect to a limited number of embodiments, it will be appreciated that many variations, modifications and other applications of the invention may be made.

Claims

WHAT IS CLAIMED IS:
1. A device for freezing a sample comprising: a. a vessel for containing the sample; b. a mechanism for moving said vessel along a track; c. a refrigeration means for imposing a laterally variable temperature gradient along said track, said refrigeration means including a plurality of thermally conductive blocks substantially enclosing said track; characterized by having a means of agitating said vessel when moving along said track.
2. The device of claim 1 wherein said vessel is a tube with a diameter of greater than about 6 mm.
3. The device of claim 1 wherein said means of agitating said vessel includes a motor.
4. The device of claim 3 wherein said means of agitating said vessel includes an electrical motor.
5. The device of claim 1 wherein said means of agitating said vessel includes a control device to vary an intensity of agitation of said vessel.
6.The device of claim 1 wherein said means of agitating said vessel are means of rotating said vessel.
7. The device of claim 6 wherein an axis of rotation of said vessel caused by said rotating means is substantially parallel to said track.
8. The device of claim 6 wherein said means of rotating said vessel includes a motor.
9. The device of claim 8 wherein said means of rotating said vessel includes an electrical motor.
10. The device of claim 6 wherein said means of rotating said vessel includes a control device to vary a rate of rotation of said vessel.
11. A method for freezing cells comprising a. confining a sample in a vessel, said vessel having a leading end and a terminal end, said sample containing the cells and having a freezing point; and b. gradually cooling said sample to below said freezing point so as to generate a temperature gradient in said sample, where during said cooling said leading end is cooler than said terminal end and to thus generate a freezing front moving from said leading end towards said terminal end; characterized in that during said gradual cooling said vessel is agitated.
12. The method of claim 11 wherein said agitation is rotation about an axis, said axis being substantially perpendicular to said freezing front.
13. The method of claim 11 wherein the sample is less than 100% of the internal volume of said vessel.
14. The method of claim 13 wherein the sample is less than about 90% of the internal volume of said vessel.
15. The method of claim 14 wherein the sample is less than about 50% of the internal volume of said vessel.
16. A method of freeze-drying cells comprising subsequent to freezing according to claim 11, said sample is lyophilized.
17. A method of storing viable freeze-dried cells comprising subsequent to said lyophilization according to claim 16, storing said sample at a storage temperature for a period of time, said storage temperature being greater than about 193K.
18. The method of claim 17. wherein said storage temperature is greater than about 253K.
19. The method of claim 18, wherein said storage temperature is greater than about 273K.
20. The method of claim 17 wherein said 'sample includes a cryoprotectant. said cryoprotectant being a solid at said storage temperature.
21. The method of 20wherein said cryoprotectant is trehalose.
22. The method of claim 17 wherein said period of time is greater than about 1 day.
23. The method of claim 22 wherein said period of time is greater than about 1 week.
24. The method of claim 23 wherein said period of time is greater than about 6 weeks.
PCT/IL2002/000738 2001-09-06 2002-09-05 Improved method for freezing viable cells WO2003020874A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/488,414 US7659111B2 (en) 2002-06-27 2002-09-05 Method for freezing viable cells
AU2002337480A AU2002337480A1 (en) 2001-09-06 2002-09-05 Improved method for freezing viable cells
EP02772760A EP1428008B1 (en) 2001-09-06 2002-09-05 Improved method for freezing viable cells
AT02772760T ATE542123T1 (en) 2002-06-27 2002-09-05 IMPROVED METHOD FOR FREEZING VIABLE CELLS

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US31714701P 2001-09-06 2001-09-06
US60/317,147 2001-09-06
US39157502P 2002-06-27 2002-06-27
US60/391,575 2002-06-27

Publications (2)

Publication Number Publication Date
WO2003020874A2 true WO2003020874A2 (en) 2003-03-13
WO2003020874A3 WO2003020874A3 (en) 2003-09-25

Family

ID=26980806

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2002/000738 WO2003020874A2 (en) 2001-09-06 2002-09-05 Improved method for freezing viable cells

Country Status (3)

Country Link
EP (1) EP1428008B1 (en)
AU (1) AU2002337480A1 (en)
WO (1) WO2003020874A2 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060099567A1 (en) * 2004-04-08 2006-05-11 Biomatrica, Inc. Integration of sample storage and sample management for life science
WO2008006621A1 (en) * 2006-07-14 2008-01-17 Rebholz, Erich Method and installation for temporarily storing microbes
US7331186B2 (en) 2002-06-27 2008-02-19 I.M.T. Interface Multigrad Technology Ltd Changing the temperature of a liquid sample and a receptacle useful therefor
US7892726B2 (en) 2004-06-07 2011-02-22 Core Dynamics Limited Method for sterilizing lyophilized eukaryotic anuclear cells with gamma irradiation
US7935478B2 (en) 2004-02-02 2011-05-03 Core Dynamics Limited Biological material and methods and solutions for preservation thereof
US20110183372A1 (en) * 2008-07-11 2011-07-28 Health Protection Agency Stimulated cell standards
WO2011098996A2 (en) 2010-02-09 2011-08-18 Core Dynamics Ltd. Improved method for changing the temperature of a biological specimen
US8037696B2 (en) 2004-08-12 2011-10-18 Core Dynamics Limited Method and apparatus for freezing or thawing of a biological material
US8196416B2 (en) 2004-02-02 2012-06-12 Core Dynamics Limited Device for directional cooling of biological matter
US8198085B2 (en) 2005-08-03 2012-06-12 Core Dynamics Limited Somatic cells for use in cell therapy
US9725703B2 (en) 2012-12-20 2017-08-08 Biomatrica, Inc. Formulations and methods for stabilizing PCR reagents
US9845489B2 (en) 2010-07-26 2017-12-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA and proteins in saliva and other biological samples during shipping and storage at ambient temperatures
US9999217B2 (en) 2010-07-26 2018-06-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA, and proteins in blood and other biological samples during shipping and storage at ambient temperatures
US10064404B2 (en) 2014-06-10 2018-09-04 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US10568317B2 (en) 2015-12-08 2020-02-25 Biomatrica, Inc. Reduction of erythrocyte sedimentation rate

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201705652D0 (en) 2017-04-07 2017-05-24 Asymptote Ltd Cryopreservation apparatus and methods

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4874690A (en) * 1988-08-26 1989-10-17 Cryopharm Corporation Lyophilization of red blood cells
US5059518A (en) * 1988-10-20 1991-10-22 Coulter Corporation Stabilized lyophilized mammalian cells and method of making same
US5071598A (en) * 1987-12-03 1991-12-10 California Institute Of Technology Cryoprotective reagent
US5364756A (en) * 1990-09-12 1994-11-15 Lifecell Method of cryopreserving a suspension of biological material
US5873254A (en) * 1996-09-06 1999-02-23 Interface Multigrad Technology Device and methods for multigradient directional cooling and warming of biological samples
US6073540A (en) * 1998-11-10 2000-06-13 Fmc Corporation Apparatus for heating or cooling product containers
JP2000189155A (en) * 1998-12-25 2000-07-11 Livestock Improvement Association Of Japan Inc Preservation of mammalian embryo or ovum and thawing dilution of frozen mammalian embryo or ovum

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU1806692C (en) * 1990-12-19 1993-04-07 Украинский Научно-Исследовательский Институт По Племенному Делу В Животноводстве Method and device for cryoconservation of farm animal semen

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5071598A (en) * 1987-12-03 1991-12-10 California Institute Of Technology Cryoprotective reagent
US4874690A (en) * 1988-08-26 1989-10-17 Cryopharm Corporation Lyophilization of red blood cells
US5059518A (en) * 1988-10-20 1991-10-22 Coulter Corporation Stabilized lyophilized mammalian cells and method of making same
US5364756A (en) * 1990-09-12 1994-11-15 Lifecell Method of cryopreserving a suspension of biological material
US5873254A (en) * 1996-09-06 1999-02-23 Interface Multigrad Technology Device and methods for multigradient directional cooling and warming of biological samples
US6073540A (en) * 1998-11-10 2000-06-13 Fmc Corporation Apparatus for heating or cooling product containers
JP2000189155A (en) * 1998-12-25 2000-07-11 Livestock Improvement Association Of Japan Inc Preservation of mammalian embryo or ovum and thawing dilution of frozen mammalian embryo or ovum

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Derwent Publications Ltd., London, GB; AN 1994-223708, XP002961154 & SU 1 806 692 A1 (ZORIN ET AL.) 07 April 1993 *
See also references of EP1428008A2 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7331186B2 (en) 2002-06-27 2008-02-19 I.M.T. Interface Multigrad Technology Ltd Changing the temperature of a liquid sample and a receptacle useful therefor
US8196416B2 (en) 2004-02-02 2012-06-12 Core Dynamics Limited Device for directional cooling of biological matter
US7935478B2 (en) 2004-02-02 2011-05-03 Core Dynamics Limited Biological material and methods and solutions for preservation thereof
US8512941B2 (en) 2004-02-02 2013-08-20 Core Dynamics Limited Biological material and methods and solutions for preservation thereof
US20060099567A1 (en) * 2004-04-08 2006-05-11 Biomatrica, Inc. Integration of sample storage and sample management for life science
US7892726B2 (en) 2004-06-07 2011-02-22 Core Dynamics Limited Method for sterilizing lyophilized eukaryotic anuclear cells with gamma irradiation
US8037696B2 (en) 2004-08-12 2011-10-18 Core Dynamics Limited Method and apparatus for freezing or thawing of a biological material
US8198085B2 (en) 2005-08-03 2012-06-12 Core Dynamics Limited Somatic cells for use in cell therapy
WO2008006621A1 (en) * 2006-07-14 2008-01-17 Rebholz, Erich Method and installation for temporarily storing microbes
US20110183372A1 (en) * 2008-07-11 2011-07-28 Health Protection Agency Stimulated cell standards
JP2011527192A (en) * 2008-07-11 2011-10-27 ヘルス プロテクション エージェンシー Stimulator cell standard
CN102144165A (en) * 2008-07-11 2011-08-03 英国健康保护局 Stimulated cell standards
US9267956B2 (en) * 2008-07-11 2016-02-23 Secretary Of State For Health Stimulated cell standards
WO2011098996A2 (en) 2010-02-09 2011-08-18 Core Dynamics Ltd. Improved method for changing the temperature of a biological specimen
US9845489B2 (en) 2010-07-26 2017-12-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA and proteins in saliva and other biological samples during shipping and storage at ambient temperatures
US9999217B2 (en) 2010-07-26 2018-06-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA, and proteins in blood and other biological samples during shipping and storage at ambient temperatures
US9725703B2 (en) 2012-12-20 2017-08-08 Biomatrica, Inc. Formulations and methods for stabilizing PCR reagents
US10064404B2 (en) 2014-06-10 2018-09-04 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US10772319B2 (en) 2014-06-10 2020-09-15 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US11672247B2 (en) 2014-06-10 2023-06-13 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US10568317B2 (en) 2015-12-08 2020-02-25 Biomatrica, Inc. Reduction of erythrocyte sedimentation rate
US11116205B2 (en) 2015-12-08 2021-09-14 Biomatrica, Inc. Reduction of erythrocyte sedimentation rate

Also Published As

Publication number Publication date
EP1428008A2 (en) 2004-06-16
AU2002337480A1 (en) 2003-03-18
WO2003020874A3 (en) 2003-09-25
EP1428008B1 (en) 2012-01-18
EP1428008A4 (en) 2004-10-13

Similar Documents

Publication Publication Date Title
US7659111B2 (en) Method for freezing viable cells
EP1428008B1 (en) Improved method for freezing viable cells
CA2051092C (en) Method and apparatus for cryopreparation, dry stabilization and rehydration of biological suspensions
JP4889602B2 (en) Storage and storage media for biological materials
Shukla Freeze drying process: A review
Gaidhani et al. Lyophilization/freeze drying–a review
CA2640590C (en) Lyophilization system and method
US5736313A (en) Method of lyophilizing platelets by incubation with high carbohydrate concentrations and supercooling prior to freezing
CA2640833C (en) Method of inducing nucleation of a material
US9642353B2 (en) Desiccated biologics and methods of preparing the same
US7153472B1 (en) Preservation and formulation of bioactive materials for storage and delivery in hydrophobic carriers
US20060019233A1 (en) Delivery of high cell mass in a syringe and related methods of cryopreserving cells
AU2001268057A1 (en) Preservation and storage medium for biological materials
WO2004011616A2 (en) Systems and methods for cell preservation
WO2006029467A1 (en) Rapid freeze drying process
Kumar et al. Fundamentals and applications of lyophilization
Xiao et al. Freeze-drying of mononuclear cells and whole blood of human cord blood
CN116694581A (en) Freeze-drying protective agent for vibrio harveyi phage V-YDF132 and preservation method thereof
Khandagale et al. Lyophilization Technique: A Review
Parthasarathy Freeze-substitution
Kendrick Preservation of fleshy fungi for taxonomy
Mazur Mechanisms of injury in frozen and frozen-dried cells
Chiotellis Lyophilization Technique for Preparing Radiopharmaceutical Kits
Meryman History and development of freeze-drying
Pandhare et al. REVIEW ON: LYOPHILIZATION PROCESS OF PHARMACEUTICALS.

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BY BZ CA CH CN CO CR CU CZ DE DM DZ EC EE ES FI GB GD GE GH HR HU ID IL IN IS JP KE KG KP KR LC LK LR LS LT LU LV MA MD MG MN MW MX MZ NO NZ OM PH PL PT RU SD SE SG SI SK SL TJ TM TN TR TZ UA UG US UZ VC VN YU ZA ZM

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ UG ZM ZW AM AZ BY KG KZ RU TJ TM AT BE BG CH CY CZ DK EE ES FI FR GB GR IE IT LU MC PT SE SK TR BF BJ CF CG CI GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 10488414

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2002772760

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002772760

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP